TO THE EDITOR:

Measurable residual disease (MRD) assessment is incorporated in the clinical decision-making of acute myeloid leukemia (AML) because of its prognostic value.1 Therefore, accurate MRD assessment is essential to ensure the appropriate consolidation therapy. Established MRD assessment includes the detection of persistent mutant NPM1 using real-time quantitative polymerase chain reaction and detection of leukemic myeloid progenitor cells using multiparameter flow cytometry. Refinement to current MRD assessments may include the detection of relapse-initiating leukemic stem cells (LSCs), defined as CD34+CD38LSC+ marker cells.2 The Dutch-Belgian Cooperative Trial Group for Hematology-Oncology (HOVON) or the Swiss Group for Clinical Cancer Research (SAKK) 102 (HO102) clinical trial and several other trials have retrospectively shown the prognostic value of CD34+CD38LSC+ burden at diagnosis3-8 and follow-up,3,9,10 in particular after 2 cycles of induction chemotherapy11 (C2). To prospectively validate the prognostic value of LSC burden, we measured the LSC burden during the HOVON-SAKK132 (HO132) randomized phase 3 trial. In this trial, the addition of lenalidomide to induction therapy was prospectively investigated in 927 patients aged between 18 and 65 years with newly diagnosed AML or high-risk MDS.12 MRD results after C2 (not LSCs) were reported to inform the consolidation treatment for the patients allocated to the 2017 European LeukemiaNet intermediate–risk group. The HO102 LSC assay was optimized and used in the HO132 trial as a standardized one-tube LSC assay13 to acquire more white blood cell (WBC) events (4 × 106).6 The LSC tube contains the backbone markers for immature myeloid progenitors (CD45, CD34, and CD38) and leukemia-specific LSC markers in the CD34+CD38 compartment (CD45RA, CD123, CD33, and CD44 and a combination of inhibitory C-lectin like receptor, T cell immunoglobulin and mucin domain 3, CD56, CD7, CD22, and CD11b in 1 fluorochrome). For details of MRD and LSC detection, we refer to our previous publications14-16 and supplemental Materials, which are available on the Blood website.

At the diagnosis, 764 of 883 (87%) eligible patients had a suitable sample, with sufficient cells for LSC detection (supplemental Figure 1). After conducting C2 in patients who achieved complete remission (CR, including CR with incomplete hematologic recovery), LSC burden could be assessed in 357 of 496 (72%) samples available for MRD measurement. The inability to assess LSC burden when the sample was available for MRD measurement was mostly because of the insufficient number of cells left after measuring MRD via multiparameter flow cytometry. No statistically significant difference in overall survival (OS) was observed between patients with or without LSC results at diagnosis and after C2 (data not shown).

Analogous to HO102 at diagnosis, 3 prognostic risk groups could be defined based on CD34 status3,17 and LSC burden. Patients were said to be with CD34AML (CD34neg) if they had <1% CD34+ blasts of WBC, no leukemia-associated–immuno-phenotype with CD34 phenotype, and no LSCs. A cut-off of 0.03% CD34+CD38LSC+ of WBC was defined in previous studies3,18 and used to distinguish LSClow and LSChigh. The LSC status at diagnosis was associated with several patient characteristics (supplemental Table 1). OS and cumulative incidence of relapse (CIR) were significantly different between the 3 groups, with the patients with CD34neg performing the best (Figure 1A,C). Prospectively assessed LSC remained an independent prognostic factor for OS and CIR after correction for WBC count at diagnosis, AML type, and ELN2017 risk group (Figure 1B). The last treatment before reaching CR/CR with incomplete hematologic recovery was also included for CIR (Figure 1D).

Figure 1.

Prognostic value of LSC burden at diagnosis. (A) Kaplan-Meier curve for OS. (C) CIR. The overall group (A,C) is divided into CD34neg, LSClow, and LSChigh, with the cut-off of 0.03% CD34+CD38LSC+ population of WBCs. (B,D) Multivariate analysis adjusted for age, AML type, ELN2017 risk group, WBCs at diagnosis, and last treatment before reaching CR (only for CIR) of (B) OS (Cox regression) and (D) CIR (Fine and Gray regression).

Figure 1.

Prognostic value of LSC burden at diagnosis. (A) Kaplan-Meier curve for OS. (C) CIR. The overall group (A,C) is divided into CD34neg, LSClow, and LSChigh, with the cut-off of 0.03% CD34+CD38LSC+ population of WBCs. (B,D) Multivariate analysis adjusted for age, AML type, ELN2017 risk group, WBCs at diagnosis, and last treatment before reaching CR (only for CIR) of (B) OS (Cox regression) and (D) CIR (Fine and Gray regression).

Close modal

LSC status after C2 was categorized into LSCneg and LSCpos based on the formerly defined 0.00000% cut-off.3 Patient characteristics based on their LSC status after C2 are summarized in supplemental Table 2. The HO132 data confirms the prognostic value of LSC in univariate and multivariate analysis of OS, (Figure 2A,B) but it did not reach statistical significance for CIR (Figure 2C,D). The combination of MRD and LSC showed that the MRDposLSCpos group had a worse prognosis compared with the MRDnegLSCneg group in univariate (supplemental Figure 2A,C) and multivariate analyses for OS and CIR (supplemental Figure 2B,D). After the sensitivity analysis, the multivariate prognostic values (Figures 1C,D and 2C,D) were corrected for the first consolidation treatment (supplemental Tables 4-6).

Figure 2.

Prognostic value of LSC burden after 2 cycles of chemotherapy. Kaplan-Meier curves of (A) OS, (C) CIR after C2. The overall group in A and C is divided into patients with LSCpos and LSCneg with the cut-off of 0.00000% CD34+CD38LSC+ population. B and D show multivariate analysis adjusted for LSC status at diagnosis, age, AML type, ELN2017 risk group, WBCs at diagnosis, and last treatment before reaching CR (only CIR) of (B) OS (Cox regression) and (D) CIR (Fine and Gray subdistribution hazard regression).

Figure 2.

Prognostic value of LSC burden after 2 cycles of chemotherapy. Kaplan-Meier curves of (A) OS, (C) CIR after C2. The overall group in A and C is divided into patients with LSCpos and LSCneg with the cut-off of 0.00000% CD34+CD38LSC+ population. B and D show multivariate analysis adjusted for LSC status at diagnosis, age, AML type, ELN2017 risk group, WBCs at diagnosis, and last treatment before reaching CR (only CIR) of (B) OS (Cox regression) and (D) CIR (Fine and Gray subdistribution hazard regression).

Close modal

In accordance with the ELN2022 recommendations for AML,1 we determined the prognostic value of LSC after conducting C2 in the different ELN2017 risk groups. For patients in the intermediate-risk group in HO132, treatment decision for an autologous (MRDneg) or allogeneic (MRDpos) stem cell transplantation (SCT) was guided by classical MRD assessment; this may explain why MRD itself was no longer a significant prognostic risk factor in this risk group.12 Remarkably, in this MRD-guided–intermediate- risk group, LSC could still be used as a marker to distinguish a group of patients with relatively poor prognosis (supplemental Figure 3E,G). The adverse-risk group comprised a relatively high percentage of patients with LSCpos (supplemental Table 2) who did significantly worse than the patients with LSCneg (supplemental Figure 3I,K). For the favorable-risk group, LSC did not have a prognostic value (supplemental Figure 3A,C), suggesting improved risk classification. The favorable-risk group as per the ELN2017 risk stratification used in the HO132 trial included less specific adverse mutations such as RUNX1 and ASXL119,20 compared with the ELN201021 risk stratification used for the HO102 protocol.

Overall, our data validate the prognostic value of LSC burden in AML at diagnosis and after C2. Interestingly, the recent publication of Li et al9 showed the prognostic value of LSC burden in AML after transplantation using a similar LSC assay. However, for the further clinical implementation of LSCs and to reach the level of standardization and harmonization that is currently established for the MRD assay, additional analyses need to be performed. For example, the use of 0.00000% cut-off for LSCs after C2 may be challenging because each positive LSC event should be specific. By acquiring a large number of WBCs in the HO132 trial (supplemental Figure 4), some (aspecific) LSC events may render the current cut-off of 0.00000% as too stringent and could potentially lead to false positives. Other optimal cut-off levels22 and/or optimization steps may be required to assess statistically significant distinct groups. Standardization and harmonization of the LSC assay, such as applicability and reproducibility was already shown at the diagnosis.14 However, to implement the LSC assay in clinical decision-making, further assay qualification experiments are warranted. In addition, the applicability of the LSC assay may be improved by including a different denominator such as CD34+ blasts or additional flow cytometric markers that can identify LSC populations in patients who are CD34.23 

Our findings on the prognostic value of LSC can have implications on clinical decision-making, for example, in cases in which LSC levels have additive value to standard MRD assessments (eg, in patients who are MRDneg; supplemental Figure 3B,D,F,H,J,L). In particular, for patients who are in the intermediate-risk MRDneg group, LSCs can play a role in refining risk group assessment, indicating a need for extensive monitoring of patients with LSCpos after autologous-SCT or even considering allogeneic-SCT for post-induction therapy. In the adverse-risk group, patients are currently allocated to allogeneic-SCT. Adaptive MRD guidance strategies have been suggested, for example, choosing different conditioning regimens; selection of donors; application of donor lymphocyte infusion or more frequent monitoring after transplantation;24 in which LSC measurement could play an additional role.25 To establish the broad implementation of LSC at different time points, extra analyses about the kinetics, subgroup analyses and the influence of therapies such as conditioning regimens and postallogeneic stem cell treatment is needed.

In conclusion, the data collected in the HO132 trial prospectively validated the prognostic value of LSCs at diagnosis and after C2 for intermediate and adverse-risk groups. Results of this study show that incorporating LSC load in clinical trials has additional value in identifying subgroups with worse prognoses.

The authors thank all participating patients and centers of the Dutch-Belgian Cooperative Trial Group for Hematology-Oncology (HOVON) and Swiss Group for Clinical Cancer Research (SAKK) 132 trial for their contribution to the study.

This research was funded by Dutch Cancer Society grant ALPE-2013-6371.

Contribution: Sample collection was done by D.A.B., T.F., B.T.G., L.G., G.J., A.A.v.d.L., J.A.M., M.G.M., T.P., J.R.P., K.P., B.L., G.J.O., and J.J.W.M.J. in the HOVON-SAKK132 trial; experiments and/or analysis of flow cytometry LSC data was performed by L.L.N., D.H., F.J., J.C.-H., L.O.-M., M.F., M.H., A.K., W.S., A.S., and P.V.; Statistical analysis was performed by L.L.N. and checked by P.G.; the manuscript was written by L.L.N. and revised by J.C., G.J.O., D.H., C.B., J.M.T., B.L., A.A.v.d.L., J.J.W.M.J., D.C.d.L., D.A.B., T.F., B.T.G., L.G., G.J., J.A.M., M.G.M., T.P., J.R.P., K.P., and P.J.M.V.; and the results were reviewed and the manuscript was approved by all the authors.

Conflict-of-interest disclosure: B.T.G. serves as a consultant for BerGenBio, Pfizer Inc, and Novartis and holds stock options in Alden Cancer Therapy and KinN Therapeutics. L.G. holds a membership on an entity’s board of directors or advisory committies for Miltenyi Biomedicine. K.P. has received honoraria for Pfizer, Novartis, Incyte, Bristol Myers Squibb, Astellas, and AbbVie and has received Celgene/Bristol Myers Squibb, Incyte, Pfizer, and Novartis. A.A.v.d.L. has received honoraria from Amgen, Novartis, Celgene/BMS, and Takeda and has received research funding from Alexion. M.GM. serves as a consultant for CDR-Life Inc; holds stock options in CDR-Life Inc; and has a patent licensed to the University of Zurich. B.L. serves as a consultant and has received honoraria from Clear Creek Bio; holds membership on an entity’s board of directors or advisory committies for Celgene, Bristol Myers Squibb, Catamaran Bio Inc, Astellas, AbbVie, F.Hoffmann La Roche. and D.C.d.L; participates in the sponsored speaker’s bureau of Servier, Roche and AbbVie; is part of the scientific advisory board of Takeda and Servier. J.J.W.M.J. serves as a consultant for Bristol Myers Squibb, Novartis, Pfizer Inc; has received research funding from Bristol-Myers Squibb, Novartis, Incyte Biosciences Benelux BV, Uppsala County Council, Glycomimetics, Avillion, and Ellipses Pharma; is a speaker bureau member for Incyte Biosciences Benelux BV, Roche, and Celgene; and holds membership on an entity's board of directors for Celgene. G.J.O. serves as a consultant for Novartis, Pfizer Inc, Celgene, Janssen, AGIOS, Amgen, Gilead, Astellas, Roche, Jazz Pharmaceuticals, and Merus; has received honoraria from Novartis, Celgene, AGIOS, Gilead, and Astellas; received research funding from Novartis; and holds membership on an entity's board of directors for Roche. J.C. serves as a consultant for Novartis; receives royalties from Navigate and BD Biosciences; and has received research funding from Takeda, DC-one, Genentech, Janssen, Novartis, and Merus. The remaining authors declare no competing financial interests.

Correspondence: Jacqueline Cloos, Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; e-mail: j.cloos@amsterdamumc.nl.

1.
Döhner
H
,
Wei
AH
,
Appelbaum
FR
, et al
.
Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN
.
Blood
.
2022
;
140
(
12
):
1345
-
1377
.
2.
Heuser
M
,
Freeman
SD
,
Ossenkoppele
GJ
, et al
.
2021 update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party
.
Blood
.
2021
;
138
(
26
):
2753
-
2767
.
3.
Zeijlemaker
W
,
Grob
T
,
Meijer
R
, et al
.
CD34(+)CD38(-) leukemic stem cell frequency to predict outcome in acute myeloid leukemia
.
Leukemia
.
2019
;
33
(
5
):
1102
-
1112
.
4.
Palmieri
R
,
Buccisano
F
,
Arena
V
, et al
.
CD34 + CD38-CLL1+ leukemic stem cells persistence measured by multiparametric flow cytometry is a biomarker of poor prognosis in adult patients with acute myeloid leukemia
.
Leuk Lymphoma
.
2022
;
63
(
4
):
996
-
1000
.
5.
Vergez
F
,
Nicolau-Travers
ML
,
Bertoli
S
, et al
.
CD34(+)CD38(-)CD123(+) leukemic stem cell frequency predicts outcome in older acute myeloid leukemia patients treated by intensive chemotherapy but not hypomethylating agents
.
Cancers
.
2020
;
12
(
5
):
1174
.
6.
Jentzsch
M
,
Bill
M
,
Nicolet
D
, et al
.
Prognostic impact of the CD34+/CD38- cell burden in patients with acute myeloid leukemia receiving allogeneic stem cell transplantation
.
Am J Hematol
.
2017
;
92
(
4
):
388
-
396
.
7.
Plesa
A
,
Dumontet
C
,
Mattei
E
, et al
.
High frequency of CD34+CD38-/low immature leukemia cells is correlated with unfavorable prognosis in acute myeloid leukemia
.
World J Stem Cells
.
2017
;
9
(
12
):
227
-
234
.
8.
Al-Mawali
A
,
Pinto
AD
,
Al-Zadjali
S
.
CD34+CD38-CD123+ cells are present in virtually all acute myeloid leukaemia blasts: a promising single unique phenotype for minimal residual disease detection
.
Acta Haematol
.
2017
;
138
(
3
):
175
-
181
.
9.
Li
S-Q
,
Xu
L-P
,
Wang
Y
, et al
.
An LSC-based MRD assay to complement the traditional MFC method for prediction of AML relapse: a prospective study
.
Blood
.
2022
;
140
(
5
):
516
-
520
.
10.
Canali
A
,
Vergnolle
I
,
Bertoli
S
, et al
.
Prognostic impact of unsupervised early assessment of bulk and leukemic stem cell measurable residual disease in acute myeloid leukemia
.
Clin Cancer Res
.
2023
;
29
(
1
):
134
-
142
.
11.
Kandeel
EZ
,
El Sharkawy
N
,
Hanafi
M
,
Samra
M
,
Kamel
A
.
Tracing leukemia stem cells and their influence on clinical course of adult acute myeloid leukemia
.
Clin Lymphoma Myeloma Leuk
.
2020
;
20
(
6
):
383
-
393
.
12.
Löwenberg
B
,
Pabst
T
,
Maertens
J
, et al
.
Addition of lenalidomide to intensive treatment in younger and middle-aged adults with newly diagnosed AML: the HOVON-SAKK-132 trial
.
Blood Adv
.
2021
;
5
(
4
):
1110
-
1121
.
13.
Zeijlemaker
W
,
Kelder
A
,
Oussoren-Brockhoff
YJM
, et al
.
A simple one-tube assay for immunophenotypical quantification of leukemic stem cells in acute myeloid leukemia
.
Leukemia
.
2016
;
30
(
2
):
439
-
446
.
14.
Hanekamp
D
,
Snel
AN
,
Kelder
A
, et al
.
Applicability and reproducibility of acute myeloid leukaemia stem cell assessment in a multi-centre setting
.
Br J Haematol
.
2020
;
190
(
6
):
891
-
900
.
15.
Cloos
J
,
Harris
JR
,
Janssen
JJWM
, et al
.
Comprehensive protocol to sample and process bone marrow for measuring measurable residual disease and leukemic stem cells in acute myeloid leukemia
.
J Vis Exp
.
2018
;
133
:
56386
.
16.
Zeijlemaker
W
,
Kelder
A
,
Cloos
J
,
Schuurhuis
GJ
.
Immunophenotypic detection of measurable residual (stem cell) disease using LAIP approach in acute myeloid leukemia
.
Curr Protoc Cytom
.
2019
;
91
(
1
):
e66
.
17.
Zeijlemaker
W
,
Kelder
A
,
Wouters
R
, et al
.
Absence of leukaemic CD34(+) cells in acute myeloid leukaemia is of high prognostic value: a longstanding controversy deciphered
.
Br J Haematol
.
2015
;
171
(
2
):
227
-
238
.
18.
Terwijn
M
,
Zeijlemaker
W
,
Kelder
A
, et al
.
Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia
.
PLoS One
.
2014
;
9
(
9
):
e107587
.
19.
Döhner
H
,
Estey
E
,
Grimwade
D
, et al
.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
.
2017
;
129
(
4
):
424
-
447
.
20.
Döhner
H
,
Estey
EH
,
Amadori
S
, et al
.
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet
.
Blood
.
2010
;
115
(
3
):
453
-
474
.
21.
Herold
T
,
Rothenberg-Thurley
M
,
Grunwald
VV
, et al
.
Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia
.
Leukemia
.
2020
;
34
(
12
):
3161
-
3172
.
22.
Ngai
LL
,
Hanekamp
D
,
Janssen
F
, et al
.
Prospective validation of CD34+CD38- leukemic stem cell frequency in the HOVON-SAKK 132 Trial: perspectives for future improvements
.
Blood
.
2022
;
140
(
Suppl 1
):
9125
-
9127
.
23.
Hanekamp
D
,
Denys
B
,
Kaspers
GJL
, et al
.
Leukaemic stem cell load at diagnosis predicts the development of relapse in young acute myeloid leukaemia patients
.
Br J Haematol
.
2018
;
183
(
3
):
512
-
516
.
24.
Czyz
A
,
Nagler
A
.
The role of measurable residual disease (MRD) in hematopoietic stem cell transplantation for hematological malignancies focusing on acute leukemia
.
Int J Mol Sci
.
2019
;
20
(
21
):
5362
.
25.
Manobianco
SA
,
Rakiewicz
T
,
Wilde
L
,
Palmisiano
ND
.
Novel mechanisms for post-transplant maintenance therapy in acute myeloid leukemia
.
Front Oncol
.
2022
;
12
:
892289
.

Author notes

Data are available on request from the corresponding author, Jacqueline Cloos (J.Cloos@amsterdamumc.nl).

The online version of this article contains a data supplement.

Sign in via your Institution